Small Vessel Disease-Related Dementia: An Invalid Neurovascular Coupling? - MDPI

Page created by Elizabeth Curtis
 
CONTINUE READING
Small Vessel Disease-Related Dementia: An Invalid Neurovascular Coupling? - MDPI
International Journal of
            Molecular Sciences

Review
Small Vessel Disease-Related Dementia: An Invalid
Neurovascular Coupling?
Rita Moretti *        and Paola Caruso
 Neurology Clinic, Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste,
 Italy; paolacaruso83@gmail.com
 * Correspondence: moretti@units.it
                                                                                                       
 Received: 11 January 2020; Accepted: 4 February 2020; Published: 7 February 2020                      

 Abstract: The arteriosclerosis-dependent alteration of brain perfusion is one of the major determinants
 in small vessel disease, since small vessels have a pivotal role in the brain’s autoregulation.
 Nevertheless, as far as we know, endothelium distress can potentiate the flow dysregulation
 and lead to subcortical vascular dementia that is related to small vessel disease (SVD), also being
 defined as subcortical vascular dementia (sVAD), as well as microglia activation, chronic hypoxia and
 hypoperfusion, vessel-tone dysregulation, altered astrocytes, and pericytes functioning blood-brain
 barrier disruption. The molecular basis of this pathology remains controversial. The apparent
 consequence (or a first event, too) is the macroscopic alteration of the neurovascular coupling. Here,
 we examined the possible mechanisms that lead a healthy aging process towards subcortical dementia.
 We remarked that SVD and white matter abnormalities related to age could be accelerated and
 potentiated by different vascular risk factors. Vascular function changes can be heavily influenced by
 genetic and epigenetic factors, which are, to the best of our knowledge, mostly unknown. Metabolic
 demands, active neurovascular coupling, correct glymphatic process, and adequate oxidative and
 inflammatory responses could be bulwarks in defense of the correct aging process; their impairments
 lead to a potentially catastrophic and non-reversible condition.

 Keywords: small vessel disease; vascular damage; endothelium; neurovascular coupling; inflammation;
 oxidative response; redox; brain’s autoregulation

1. Introduction
      Cerebral small vessel disease (SVD) primarily distresses the small perforating arteries, being
defined as vessels with less than 50 µm diameters, also defined as “all the vessels within the brain
parenchyma plus the vessels with a diameter less than 500 µm in the leptomeningeal space” supplying
the deep brain structures [1,2]. Nevertheless, general increased arterial stiffness is associated with an
increased white matter lesion burden [3]. Therefore, while the microvasculature is the primary target
of SVD, the contribution of larger arteries should not be immediately discounted. SVD is the most
important and common cause of vascular dementia, leading to 45% of dementia, and it accounts for
about 20–30% of all strokes worldwide, 25% of ischemic (or lacunar strokes). Moreover, it significantly
increases the risk of future stroke [4]. Often, SVD lesions are clinically insidious and they act as “silent”
lesions. Thus, SVD is a dynamic pathology, lesions progress over time, and the long-term outcome and
impact on brain damage vary [5]. In sporadic cerebral SVD, sporadic aging and hypertension are listed
as the most critical risk factors. However, different hereditary forms of cerebral SVD have also been
described [6]. In the latter forms, several pathological changes to the vasculature in small arterioles
(like vascular muscle dysfunction, lipohyalinosis, vascular remodeling, and the deposition of fibrotic
material) have been identified. Venous structures are also affected [7]. These facts are shared in both
forms, with time of onset beng the only difference.

Int. J. Mol. Sci. 2020, 21, 1095; doi:10.3390/ijms21031095                           www.mdpi.com/journal/ijms
Small Vessel Disease-Related Dementia: An Invalid Neurovascular Coupling? - MDPI
Int. J. Mol. Sci. 2020, 21, 1095                                                                        2 of 35

     Cerebral amyloid angiopathy (CAA) is a common form of cerebral SVD and it refers to the
deposition of amyloid b-peptide (Ab) in the cerebral leptomeningeal and parenchymal arteries and
arterioles walls. The incidence of CAA increases with age. More often, CAA is related to hemorrhagic
stroke. Additionally, in this case cause, structural variations, such as concentric splitting, loss of smooth
muscle cells, and fibrinoid necrosis, which may increase the propensity for vessel rupture and, thus,
hemorrhage, have been seen [8,9].

2. Vascular Dementia and Small Vessel Disease-Related Dementia
      The diagnosis of vascular dementia should be easy due to the temporal correlation between an
acute vascular brain lesion and the onset of cognitive and behavioral problems. Nonetheless, consensus
criteria for vascular cognitive impairment are still under debate, since 1983, when NINDS-AIREN
Criteria had first been written [10,11], and the ICD-10 had been debated [12]. Subsequently, different
validations have been proposed, and many criteria have been written, but the current clinical diagnostic
criteria for vascular dementia are still argued, even from a neuropathological perspective. Nowadays,
we accept the generic definition of genetic vascular dementia (CADASIL or CARASIL), macrovascular
dementia (multi-infarct dementia or strategic infarct dementia), or microvascular dementia (subcortical
vascular dementia or more appropriately, small vessel disease-related dementia) [13–15]. Very recently,
DSM V [16,17] and the STRIVE Consortium (Standards for reporting Vascular changes on Neuroimaging)
conditioned the diagnostic criteria to specific neuroimaging studies [5,18]. In particular, the diagnostic
criteria for the small vessel disease should include, in a conventional MRI, recent subcortical infarcts,
white matter hyperintensities, lacunes, prominent perivascular spaces, and cerebral microbleeds [5,18].
Therefore, we take small vessel disease (SVD) into account, which is the consequence of the different
damages to the small penetrating arteries and arterioles in the pial and lepto-meningeal circulation,
along with penetrating and parenchymal arteries and arterioles, pericytes, capillaries, and venules [19].
SVD prevalence increases exponentially with aging. Around Europe, the prevalence rates of SVD related
dementia, between ages 65–69 to 80+ years, ranged from 2.2 to 16.3% [20–23]. As aforesaid, aging is the
most critical risk factor in developing the small-vessel disease, due to the loss of arterial elasticity, and a
consequent reduction of arterial compliance [24]. The loss of arterial compliance is the major determinant
of the altered autoregulation capacities, which leads to the deep sensitiveness of the brain of SVD
patients to brisk decreases of blood pressure [25–27]. Moreover, apart from the reduction of elasticity, it
should be considered that aging also causes a low-level functioning of the autonomic nervous system,
with direct and endothelium-mediated altered baroreflex activity [28–31]. Pathological expressions of
SVD are the arteriolosclerosis process and cerebral amyloid angiopathy (CAA) [32–35]. After that, even
if debated, SVD could affect the integrity of the medial cholinergic pathway, for the hypoperfusion
preferred localization, in the deep white matter capsule, [36], or, due to the multiple lacunar infarcts,
the basal forebrain cholinergic bundle could be deafferentated from the tubero-mamillary tracts [37,38].
These aspects affect the normally-accurate cerebral flow regulation and they can further disturb
the “retrograde vasodilatation system” with necessary consequences in neurovascular coupling [39].
Cerebral small vessel disease includes a neuroimaging and pathological descriptions, which comprise
different imaging changes in the white matter and subcortical grey matter, including small subcortical
infarct, lacunes, white matter hyperintensities (WMHs), prominent perivascular spaces (PVS), cerebral
microbleeds (CMBs), and atrophy. Moreover, an associated hypoperfusion progression characterized
SVD, causing incomplete ischemia of the deep white matter [7,40–42] accompanied by inflammation,
diffuse rarefaction of myelin sheaths, axonal disruption, and astrocyte gliosis [35]. In small vessel
disease, the occlusion of deep periventricular-draining veins is also evident [43], together with a
disruption of the blood-brain barrier; the two factors together causing a severe leakage of fluid and
plasma cells to potentiate the inflammatory cascade, which seem to happen in the course of chronic
hypoperfusion, by collecting multifactorial causes for white matter alterations [44–46]. Cerebral small
vessel disease is what has been described as “a progressive disease” [35]. Lesions progress over
time, and the long-term outcome and impact on brain damage vary, even not knowing why or how;
Small Vessel Disease-Related Dementia: An Invalid Neurovascular Coupling? - MDPI
Int. J. Mol. Sci. 2020, 21, 1095                                                                                              3 of 35

 been described as “a progressive disease” [35]. Lesions progress over time, and the long-term
Int. J. Mol. Sci.
 outcome      and 2020, 21, 1095on
                     impact      brain damage vary, even not knowing why or how; reasonably, it should                         3 of 35
                                                                                                                                     be
 said that the most rapid and confluent progression of the isolated white matter hyperintensities could
 be considered
reasonably,          as being
                it should       the most
                            be said         relevant,
                                     that the          to theand
                                               most rapid      best   of knowledge,
                                                                   confluent              predictor
                                                                                progression     of theofisolated
                                                                                                          the fatalwhite
                                                                                                                      progression
                                                                                                                             matter
 of  SVD     [47–50].  Of   course,  the  total amount    of  lacunes     and  profound      white
hyperintensities could be considered as being the most relevant, to the best of knowledge, predictor matter    alterations      relate
oftothe
     thefatal
           degree    of cognitive
                progression         impairment
                                of SVD    [47–50]. [51–53].
                                                    Of course, Thethepreferred     location
                                                                        total amount          of the lesions
                                                                                          of lacunes             is placedwhite
                                                                                                        and profound            along
 with    the  frontal  and    prefrontal-thalamus-basal       forebrain     networks,     [54,55],  directly
matter alterations relate to the degree of cognitive impairment [51–53]. The preferred location of the         implying       the   so-
 called    cortical-deafferentation.       Additionally,   lesions    due   to  SVD    are  specific
lesions is placed along with the frontal and prefrontal-thalamus-basal forebrain networks, [54,55],   to   the caudate      nucleus
 (the most
directly        precociously
           implying             affected
                        the so-called      region), the putamen, Additionally,
                                        cortical-deafferentation.      insula, precentral      gyrus,
                                                                                         lesions  due toinferior
                                                                                                            SVD are frontal    gyrus,
                                                                                                                        specific    to
 and   middle      frontal  gyrus.  The    higher  metabolic     request    of these   regions   (more
the caudate nucleus (the most precociously affected region), the putamen, insula, precentral gyrus,        than   20%)     at steady
 state than
inferior         other
           frontal      brainand
                     gyrus,     areas  fullyfrontal
                                   middle     explains   the pathology
                                                     gyrus.    The higher [56–63].
                                                                              metabolic  Onrequest
                                                                                              the other     hand,
                                                                                                      of these       SVD usually
                                                                                                                  regions     (more
than 20%) at steady state than other brain areas fully explains the pathology [56–63]. On the[64,65];
 implies      a  reduced     metabolic    rate  of  oxygen     (estimated      of  about    35%    in  white    matter)        other
 metabolic
hand,     SVD incongruity
                 usually impliesbetween    the brain
                                      a reduced        oxygen rate
                                                   metabolic      supply    and its consumption
                                                                       of oxygen      (estimated of has  aboutbeen35% described
                                                                                                                          in whitein
 SVD, which
matter)            determines
            [64,65];  metabolicanincongruity
                                      altered neurovascular
                                                   between thecoupling         and altered
                                                                     brain oxygen       supply vasomotor      reactivity [35,66–
                                                                                                  and its consumption            has
 71].  Neuropsychological          pattern    profiles  of  dementia       that  are
been described in SVD, which determines an altered neurovascular coupling and altered  related   to  SVD     are   related     to the
                                                                                                                       vasomotor
 subcortical-cortical
reactivity     [35,66–71].loops    deafferentation and
                              Neuropsychological           they are
                                                        pattern        distinguished
                                                                   profiles   of dementia by poor
                                                                                               that executive
                                                                                                     are related   function,
                                                                                                                      to SVD poor are
 planning,      working     memory      alterations,  loss  of  inhibition,    reduced     mental    flexibility,
related to the subcortical-cortical loops deafferentation and they are distinguished by poor executive               multitasking
 procedures
function,     poorinvalidation,   and decrease
                     planning, working       memoryspeed   of executive
                                                        alterations,        process
                                                                         loss          [72–78].reduced
                                                                               of inhibition,     Any specific
                                                                                                             mental treatment      has
                                                                                                                        flexibility,
 been discovered,
multitasking             either as
                   procedures       pathogenicand
                                 invalidation,     or highly
                                                       decreasestandard
                                                                    speed recommended
                                                                            of executive processfor this   condition.
                                                                                                       [72–78].     Any specific
        Insert   Figure   1  appr. here:
treatment has been discovered, either as pathogenic or highly standard recommended for this condition.
      Insert Figure 1 appr. here:

         Figure 1. A synopsis of the possible superimposing factors conditioning the progression of SVD.
          Figure 1. A synopsis of the possible superimposing factors conditioning the progression of SVD.
3. Anatomical and Structural Weaknesses in Small Vessel Disease
 3. Anatomical     and Structural
     SVD is considered                Weaknesses
                              to be the               in Small Vessel
                                         major contributing                Disease
                                                                   factor or the sole responsible for the “generic
defined”   dementia-syndrome        worldwide     [79].  The    small
        SVD is considered to be the major contributing factor or the   vessels represent   its principal
                                                                                   sole responsible    fortarget, which
                                                                                                            the “generic
include
 defined” pial and small penetrating
             dementia-syndrome             arteries,[79].
                                      worldwide      smallThe intra-parenchymal     arterioles
                                                                 small vessels represent        (with smooth
                                                                                            its principal       muscle
                                                                                                           target, which
cells),  perivascular   spaces,   astrocytic  endfeet,   cerebral    capillaries and   veins,  and
 include pial and small penetrating arteries, small intra-parenchymal arterioles (with smooth musclevenules.   There  is
wide
 cells),speculation
          perivascularon spaces,
                          all the structures
                                   astrocytic involved,      to establish
                                               endfeet, cerebral           a potential
                                                                      capillaries        role inand
                                                                                   and veins,     the venules.
                                                                                                      development
                                                                                                                Thereofis
the chronic   ischemic-hypoxic      state, which   is the  final  responsibility  for the  SVD,   even if
 wide speculation on all the structures involved, to establish a potential role in the development of the the principal
SVD-model     is the arterioles damage-based,
 chronic ischemic-hypoxic        state, which isand the even
                                                         finalifresponsibility
                                                                  we do not know formuch    abouteven
                                                                                      the SVD,     perivascular   spaces.
                                                                                                        if the principal
The  pathophysiological
 SVD-model                   role ofdamage-based,
                is the arterioles    PVS, their functionand andeveninteraction  withknow
                                                                     if we do not      cerebral
                                                                                             muchmicrocirculation,   has
                                                                                                    about perivascular
not been established yet. There is a broad consensus that PVS forms a network of spaces around
cerebral microvessels, acting as a canal for fluid transport, the exchange between cerebrospinal fluid
Int. J. Mol. Sci. 2020, 21, 1095                                                                      4 of 35

(CSF), and interstitial fluid (ISF) and the clearance of catabolites from the brain. The perivascular
compartment contains several cell types, like perivascular macrophages, pial cells, mast cells, nerve
fibers, and collagen fibers [80]. Usually, as arterioles penetrate deeper into the brain, the glial
membrane, the pericyte membrane fuse together and then obliterate the perivascular spaces [80,81],
but it has been proposed that either in humans either in animals, the perivascular space could act as a
brain lymphatic system, also being defined as “para-arteriolar”, “para-venular”, “paravascular”, or
“glymphatic” [82]. This system has many complex functions (further in the review, we will explain
it regarding neurovascular coupling), but it seems likely to exert the drainage work of the brain.
Therefore, modification of this system produces deleterious effects, whose results are an accumulation
of catabolites and toxic substances, together with a pronounced neural starvation [83,84]. In SVD,
this system is invalid; one of the SVD hallmarks is the enlargement and widening of PVS, due to an
obstructive process that is maintained by catabolites, proteins, and cell debris [82]. In small vessel
disease, the occlusion of deep periventricular-draining veins is also evident [43], together with the
disruption of the blood-brain barrier (BBB). All these facts together lead a consequent leakage of fluid
and plasma cells, which eventually might potentiate the perivascular inflammation, and all of the
cascades of the inflammatory/obstructive/stagnation-induced process [44–46,85]. The immobility of
the fluid drainage can support PVS’s role in different diseases: the possible explanation of the PVS
involvement in SVD, is the argued relationship demonstrated between an altered cerebrovascular
reactivity (CVR), which is the change in cerebral blood flow in response to a vaso-active stimulus in
the so-called neurovascular coupling, the found BBB dysfunction, and the correspondent perivascular
inflammation [86]. Therefore, a lacuna should not indicate an enlarged perivascular space, as it is, still
nowdays; it should never be the correspondent of the CSF-filled cavities on brain MRI or residual
lesion of a small hemorrhage [82,87–92]. Nowadays, it should be more appropriate for the definition
“lacuna of presumed vascular origin” to replace the term ”lacuna” [20,93–96].

3.1. Arteriolosclerosis as a Functional Model for SVD
      Arterioles are the best studied target for SVD, starting from the pathological process that they
undergo, the arteriolosclerosis. Arteriolosclerosis occurs in two primary histological forms, the
hyperplastic and the hyaline arteriolosclerosis [97,98]. The hyperplastic is the most common lesion,
principally due to the chronic state of hypertension. It begins with the hypertrophy of the smooth
muscle in the media, and it is accompanied by the reduplication of elastic laminae, the growth of new
cells in the intima, and the deposition of collagen, which progressively substitutes the muscle cells
(onion skin arteries) and severely obliterates the lumen [97]. Hyaline sclerosis is another change in
the vessels of hypertensive patients: the vessel wall becomes thickened with collagen [99]. Arterioles
undergo a progressive deposition of hyaline material throughout the entire circumference of the vessel
and which extends through the media [100]. The hyaline material is a consequence of the leakage of the
plasma proteins, mainly the inactive form of complement (C3b) through the endothelium, and also by
an increment of the basement membrane components by the smooth muscle cells [100]. Healthy aging
implies the loss of the Windkessel effect and the loss of arterial elasticity, which reflects an anticipated
and precocious return of the so-called wave reflection. Healthy aging also determines an increase of
the systolic and a decrease of diastolic pressure, with a loss of resting flow effect through the Willis,
which decrements the usual high perfusion pressure towards the most profound small arteries of the
brain [101–103], thus provoking a loss of brain flow autoregulation. Arteriolosclerosis perpetuates the
hypo-perfusion in the profound territories that are irrigated by penetrating arteries.

3.2. Hypoperfusion and Neuroinflammation
     It is intriguing enough that chronic cerebral hypoperfusion defeats the traditional and acknowledged
way of the anatomical thinking, with regards to the preponderance of cortical neurons on the other
brain structures. Nowadays, it is well accepted that 10 min. of transient global ischemia in rat-brains
determines a precocious sufferance of the perineural spaces, and then of the white matter, along with
Int. J. Mol. Sci. 2020, 21, 1095                                                                      5 of 35

the internal and external capsule; one day after the ischemia, oligodendrocytes die [104–106], and the
neuronal death occurs several days after the initial damage [107,108]. Moreover, as experimentally
demonstrated, ischemia occurs in the brain (rat, mouse, and rabbit) [109,110], it seems evident that
there is an induction of significant microglial activation, with significant regional variability [109].
When measured the time of onset, it has been described that microglial activation firstly appears in
the hippocampus, but the activation does not last more than 48 h [109]. In the meantime, from 48
to 72 h after the ischemia, there is increased activation of the microglia. It occurs throughout the
white matter, and the thalamus (from the second day after up to the fourth day). From the fourth
day, the activation occurs through the cortex, protracting until 30 days after the initial ischemia [111].
Besides, microglia tend to retract their branches after ischemia, leading to a reduction in the total length
and the total number of microglial processes [112]. The loss of blood flow in the peri-infarct region
results in marked de-ramification and amoeboid transformation of soma [113]. Microglial activation is
believed to be involved in the pathological progression of ischemic tissue. However, the function of
activated microglia in ischemic events remains not entirely understood [113–115]. The experimental
models seem to validate the hypothesis of two-step sequential microglia activation: the first one,
mostly dependent on M1 type activation, with the production of oxidative species, proinflammatory
cytokines, and lysosomal cascades [116]; soon after, there is an M2 activation, which seems to be
reparative and blocking the inflammatory cascade of events [113,116]. It has been supposed that
when the ischemic event is not an acute one, but there is chronic ischemia, like in SVD, there is a
preponderance of M1 activation, with minimal M2 action [117–120]. Chronic ischemia determines
a severe oligodendrocyte degeneration; soon after, it causes microglial activation and it is further
associated with an increase of apoptosis processes that are associated with an elevation of caspase
3 RNA, and of matrix-metalloprotease 2 (MMP-2) expression [121,122]. Astrocytes react to the chronic
ischemic condition, as a result of the length and severity of the insult. In the early ischemic period,
the astrocytes respond with a remarkable proliferation, but, in the case of persistent hypoperfusion,
with their degeneration and death [123–125]. It has been argued that astrocytes act in response to
the ongoing modification of the neurons metabolic changed requests, possibly through glutamate
signaling [126], and contribute to the regulation of blood flow modifying capillary permeability, by
stretching out their endfeet to the microvessel, establishing a proximal connection with the capillary.
Their death, due to chronic hypoperfusion, leads to an expanding, and auto-potentiating system of
neuronal death, due to a misleading neurovascular coupling. The deterioration of astrocytic function
at the late stages of white matter hyperintensities also supports the progressive character of SVD, as
shown in a recent clinicopathological study [127]. The more actual histological works discovered
collagenous pouches and tubes around small vessels, now referred to as vascular bagging, suggested as
a possible biological marker of SVD [128]. Ultrastructural studies have found the splitting, branching,
and thickening of the capillary basement membrane and perivascular deposition of collagen, also called
microvascular fibrosis, in the brains of aged rats [129] and rhesus monkeys [130]. Frosberg et al. [128]
showed vascular bagging in the frontoparietal and temporal control deep white matter. The Authors
found that plasma proteins fill the vascular bagging, and argued that SVD should be characterized
by a porous endothelium and an altered basement membrane [128,131]. Frosberg et al. [128] showed
that, in SVD with diffuse white matter alteration, smaller basal ganglia vessels, including pre-capillary
arterioles and capillaries, revealed vascular bags with COLL4-positive walls [132–134]. Post-capillary
venules also showed vascular bagging in SVD, but they cannot be distinguished from capillaries,
merely based on vessel diameters, deformed erythrocytes squeezing through vessels, or the presence
of pericytes [135]. While the pericytes that were found in the Frosberg et al. work [128] were located
outside the vascular bags, these cells and their processes are enclosed by two layers of the basement
membrane [130], and therefore their degeneration might contribute to a splitting of the basement
membrane, supporting what we have afore-described. Frosberg et al. [128] also reported the presence
of string vessels in SVD. String vessels are thin connective tissue strands, remnants of capillaries, with
no endothelial cells and without the primary function of blood transport [129]. String vessels suggest
Int. J. Mol. Sci. 2020, 21, 1095                                                                     6 of 35

the precise location of the originally normal-functioning vessels, and after significant events (abrupt
or chronic ischemia, aging, but also neurodegenerative disorders), they gradually disappear [136].
Many events induce their regression, which is probably due to a converging two-vias: an induced
apoptotic phenomenon associated with the destruction of endothelial cells, attached by macrophages.
Frequently the regression might be triggered by the loss of the vascular endothelium grogth factor
(VEGF) [136]. In their work, Frosberg et al. [128] put in evidence four types of string vessels, suggesting
different stages of string vessel formation, and an enhanced density of COLL4-positive string vessels
and ghost vessels that resembled remnants of string vessels [128]. Quite suggestive is the finding that
the higher quantities of string vessels that are described in work [128] have been found in the damaged
white matter parenchyma. However, in some cases, the Authors have found that after an endothelial
death, the empty basement membrane tubes could help the regrowth of new endothelial cells, which
can synthesize new basement membrane layers [137], which could give the reason of the multi-layered
vascular bags that are found in the original work [128].

3.3. Cholinergic Role in Small Vessel Disease
      Moreover, small arteries undergo a systemic poorness of cholinergic network regulation.
Many hypotheses have been raised for a possible explanation, starting from an altered cholinergic
response to inflammation, which is a constant in chronic ischemic condition [138–141], up to a
disruption of the cholinergic networks, which subcortically approaches the basal forebrain, since this is
a preferential location of lacunar vascular infarcts and chronic hypoperfusion syndrome [142–145].
It has been widely demonstrated, either in animal models either in postmortem studies, that there
is a reduced level of acetylcholine (Ach) in patients with vascular dementia [146,147], both in the
cortical areas, in the hippocampus, and the cerebrospinal fluid [148–151]. A loss of cholinergic
neurons in 40% of demented vascular patients was reported, accompanied by reduced ACh activity
in the cortex, hippocampus, and striatum [152]. Post-mortem SVD studies revealed lower choline
acetyltransferase (ChAT) activity when compared with the controls [153], and SVD patients have
lower CSF concentrations of Ach [151,154–156]. In fact, in the experimental condition, the selective
muscarinic antagonism by atropine, for example, has dramatic consequences in the CA1 region, [157].
Other experiments demonstrated that the selective alpha-7-nicotinic AChR antagonism exacerbates
the hypoxic effects on the CA1 and CA3 cortical areas; on the contrary, non-selective nicotinic AChR
antagonists have a detrimental effect on the hippocampus, not in all the other cortical areas [158–161].
The chronic reduction of the cerebral blood flow can affect the control of the cholinergic networks, but
it happens that a proper cholinergic function is compulsory to well-regulation of the regional brain
blood flow [162,163]. Ach principally mediates the parasympathetic innervation of the Willis circle and
the pial vessels [164]. Ach stimulates in vitro the arterial relaxation, directly and via the promotion of
the synthesis of vasodilator endothelium agents [164], via the nitric oxide synthase [165] and the GABA
interneurons [166–168]. The stimulation of the Nucleus Basalis of Meynert results in increased blood
flow throughout the cerebral cortex in experimental animals [169]. Upon stimulation, perivascular
cortical afferents release Ach into endothelial M5 muscarinic receptors [148,170]. M5 receptors are
highly expressed in blood vessel walls [170]. Yamada et al. [148] prepared knockout mice (M5−/−) and
found that, as compared to wild-type mice, these animals lose the ability to dilate cerebral arteries, but
could still regulate extra-cerebral flow. Upon stimulation, perivascular cortical afferents release Ach
onto endothelial M5 muscarinic receptors [170,171]. Hamner et al. [172] demonstrated that cholinergic
control of the cerebral vasculature might be active at low frequencies, lower than 0.05 Hz when the
sympathetic nervous system appears to play a role in the cerebral auto-regulation, in a limited but
well-conducted study. At these low frequencies, the myogenic mechanisms appear to play any role; to
surprise, the correspondence between cholinergic and sympathetic cerebrovascular regulation above
0.05 Hz is striking, suggesting that the cerebral circulation engages different mechanisms to protect
itself [172]. There are many different reasons for the cholinergic impairment that was observed in
SVD. The cholinergic impairment for artery dysregulation that was observed in small vessel disease
Int. J. Mol. Sci. 2020, 21, 1095                                                                   7 of 35

could derive from the deafferentation of the basal forebrain cholinergic bundle to the subcortical
structures, due to the most probable location of lacunar vascular events and the chronic hypoperfusion
consequences, aforementioned [36,38,173]. Bohnen et al. [36] demonstrated in vivo that the severity of
the periventricular white matter lesions is associated with lower AchE activity, in the middle-aged
and elderly subjects without dementia, as a result of cortical cholinergic deafferentation. In animal
SVD models, there is a concomitant reduction of vasopressin and histamine, which is interpreted as a
result of the interruption of the tracts that comes from the supra-optic and tuberomammillary nuclei
and ends in the basal forebrain [174]. The reduction of vasopressin and histamine seems to have a
redundant effect on hypoperfusion. Some clinical data confirm a reduction of the number of cholinergic
neurons in the Nucleus Basalis of Meynert in multi-infarct dementia, but not in SVD [175–177]. It has
been conveyed that a primary loss of the cholinergic neurons of Nucleus Basalis of Meynert does
not mediate cholinergic impairment [178,179], but is a consequence of the secondary cholinergic
deficits, due to the indirect, cholinergic endothelial effect, aforementioned. Though, the number of
muscarinic cholinergic receptors is markedly reduced in mixed dementia patients [179] and SVD
dementia. Cholinergic poorness promotes a less efficacious endothelium relaxation, even due to
an altered nitric oxide synthase and loss of efficacy of the GABA interneurons [165,166]. The two
mediators seem to be less efficient in influencing the small arteries contraction [180–183]. A final step
on this point has been written by a probabilistic tractography analysis [155]; this study tracked the
two primary white matter tracks which map to cholinergic pathways, identifying a significantly lower
fractional anisotropy in precocious form of SVD. Mediation analysis demonstrated that fractional
anisotropy in the tracked pathways could fully account for the executive dysfunction, and partly
mediate the memory and global cognition impairment. The recently published study [155] study
suggests that the fibers that are mapped into the cholinergic pathways, but not those of the Nucleus
Basalis of Meynert, are significantly damaged.
     Finally, an alteration of the conceptualized “cholinergic anti-inflammation pathway” summarized
another possible cause for the cholinergic poorness [138,139]; these findings are based on the
knowledge that acetylcholine released from cholinergic axon terminals can interact with α7 nicotinic
Ach receptors on vicinal immune cells. The nicotinic receptors then translate the cholinergic signal into
the suppression of cytokine release, being involved in the inflammatory cascade [140,141]. A chronic
proinflammatory condition counterbalances the acetylcholine release and promotes its cascade effects
on the vasoregulation. The pathological cascade of events, which occurs as a consequence of all
the pathological alterations described, determines a decrease of the vascular tone, with a release of
the blood-brain barrier permeability, with a loss of the internal vascular remodeling and with major
vascular rarefactions. As a result, hypo-perfusion at rest occurs in the brain and it is associated with
impairment in the moment-to-moment control of CBF, with a decrease of adaptive vascular responses
and with a diminishment of the neurovascular coupling and auto-regulation system [145,180].
     Insert Figure 2 approx here:
Int.Int.
     J. Mol. Sci.Sci.
         J. Mol.  2020, 21,21,
                      2020, 1095
                               1095                                                                                             8 of
                                                                                                                                   8 35
                                                                                                                                     of 35

                               Figure 2. Confirmed pathological processes underlying SVD.
                                  Figure 2. Confirmed pathological processes underlying SVD.
4. Chronic Hypoxia and Brain Response
   4. Chronic Hypoxia and Brain Response
      The brain requires a disproportionate amount of the body’s energy. Up to 20% of cardiac output
         Thetobrain
is devoted             requires
                 meeting             a disproportionate
                              the brain’s    energy demands, amount       of theaccounting
                                                                    despite       body’s energy.
                                                                                              for only Up2% to 20%    of cardiac
                                                                                                                of body            output
                                                                                                                           mass [184].
   is devoted    to  meeting      the  brain’s  energy   demands,        despite   accounting
The cerebral vasculature possesses well-developed mechanisms that enable cerebral blood flow (CBF) for  only  2%    of body   mass   [184].
   The   cerebral     vasculature       possesses     well-developed          mechanisms       that
to remain constant during fluctuations in arterial pressure (autoregulation) and meet the increased    enable     cerebral    blood   flow
   (CBF) demands
nutrient    to remainwhen   constant
                                   localduring     fluctuations
                                          brain activity     rises inin order
                                                                           arterial  pressurethese
                                                                                 to deliver       (autoregulation)        and meet
                                                                                                        nutrients effectively       andthe
   increased     nutrient       demands      when     local   brain    activity    rises  in
protect the brain from hypoperfusion and ischemic damage [185,186]. Cerebral SVD significantlyorder      to  deliver    these   nutrients
andeffectively
      chronicallyand    protectthe
                     impairs        theability
                                        brain offrom
                                                   thehypoperfusion
                                                        cerebral vasculature  and ischemic
                                                                                     to meet damage         [185,186].
                                                                                                these demands          dueCerebral
                                                                                                                            to severalSVD
   significantly     and    chronically     impairs    the  ability    of  the  cerebral   vasculature
structural and functional changes, which ultimately result in brain injury, cognitive decline, and           to  meet    these  demands
   due to several
dementia.     Cerebral structural      and functional
                            vasoconstrictor                changes, responses
                                                 and vasodilator          which ultimately       result in
                                                                                      are important           brain injury,
                                                                                                           mechanisms        by cognitive
                                                                                                                                 which
   decline,
brain           and isdementia.
       blood flow         maintained,    Cerebral      vasoconstrictor
                                           as aforementioned.        In SVD,   and    vasodilator
                                                                                 chronic   hypoperfusion responses
                                                                                                                 leads toarea decrease
                                                                                                                               important
   mechanisms
in cerebral   bloodby flow, which
                               hypoxia,brain   blood stress,
                                           oxidative    flow and is maintained,        as aforementioned.
                                                                       triggers inflammatory         responses, which In SVD,leadschronic
                                                                                                                                    to a
   hypoperfusion         leads    to  a decrease     in cerebral    blood      flow,  hypoxia,
potentiated hypoperfusion condition. The induced lesions are mostly expressed in the white matter   oxidative      stress,  and   triggers
   inflammatory
(WM)    and especiallyresponses,       which leads to aWM,
                             in the periventricular          potentiated      hypoperfusion
                                                                  basal ganglia,                   condition. Hypoxia-induced
                                                                                      and hippocampus.            The induced lesions
oxidative stress leads then to mitochondrial dysfunction, neuronal damage, and apoptosisWM,
   are  mostly     expressed       in  the  white    matter    (WM)       and   especially   in   the   periventricular              basal
                                                                                                                                via the
   ganglia,
nitric oxide and       hippocampus.
                synthase       (NOS) pathway  Hypoxia-induced
                                                     [187–190]. Chronic   oxidative    stress
                                                                                 hypoxia          leads then
                                                                                            profoundly               to mitochondrial
                                                                                                               influences     vascular
control, altering both vasoconstrictors as well as vasodilator responses in isolated cerebral [187–190].
   dysfunction,      neuronal      damage,     and  apoptosis     via   the  nitric oxide  synthase       (NOS)    pathway     vessels;
   Chronic
indeed,        hypoxia
          chronic    hypoxiaprofoundly
                                   alters theinfluences
                                               contractile vascular
                                                              response    control,  altering cerebral
                                                                            of the isolated    both vasoconstrictors           as well as
                                                                                                            vessels [191]. Chronic
   vasodilator
hypoxia    is known responses       in isolated
                         to influence              cerebral
                                          Nitric Oxide      (NO)vessels;     indeed,ofchronic
                                                                    modulation          contractilehypoxia      altersInthe
                                                                                                         response.        onecontractile
                                                                                                                                animal
   response     of the    isolated    cerebral   vessels   [191].   Chronic      hypoxia   is  known
study, the authors showed that chronic hypoxia augmented contractile sensitivity to the thromboxane        to influence     Nitric  Oxide
   (NO)    modulation         of  contractile    response.     In  one     animal   study,   the    authors
mimetic U-46619 in isolated cerebral vessels as the result of reduced nitric oxide (NO) production and          showed      that  chronic
   hypoxia
activity.      augmented
          A decrease       in NO contractile
                                     productionsensitivity    to theand
                                                   of L-arginine       thromboxane       mimetic
                                                                             oxygen increased       NO U-46619     in isolated
                                                                                                           degradation           cerebral
                                                                                                                           or reduced
   vessels    as  the   result     of  reduced     nitric  oxide    (NO)       production
cyclic guanosine-3-5-monophosphate (cGMP) production (involved in smooth muscle relaxation).  and     activity.    A   decrease    in NO
In production
    this case, the of administration
                       L-arginine and of      oxygen    increased NO
                                                 the nonspecific         NO degradation
                                                                               synthase (NOS) or reduced
                                                                                                      inhibitor cyclic   guanosine-3-5-
                                                                                                                    nitro-L-arginine
   monophosphate            (cGMP)      production      (involved        in  smooth    muscle
(NLA) eliminated the difference in contractile sensitivity between the vessels from the normoxic    relaxation).      In  this case,andthe
   administration       of   the   nonspecific    NO    synthase      (NOS)     inhibitor   nitro-L-arginine
chronically hypoxic animals, which suggests that a reduction in NO production and activity was                       (NLA)    eliminated
   the difference
responsible     for the in increased
                             contractilecontractile
                                             sensitivitysensitivity
                                                           between that   the vessels    from the
                                                                                was observed             normoxic
                                                                                                    [192].             and chronically
                                                                                                              Such effects     may be
   hypoxic animals, which suggests that a reduction in NO production and activity was responsible for
   the increased contractile sensitivity that was observed [192]. Such effects may be significant in the
Int. J. Mol. Sci. 2020, 21, 1095                                                                   9 of 35

significant in the adult in whom disorders involving cerebral circulation occur under conditions of
acute and chronic hypoxia. Chronic cerebral hypoperfusion (CCH) is a prevalent pathophysiological
state in patients with Alzheimer’s disease (AD) and vascular dementia (VaD). CCH has been identified
as one of the initial conditions that are critical in the development of cognitive dysfunction [193].
In several studies, deranged energy metabolism, glial activation, apoptosis, oxidative stress, neuronal
damage, and white matter lesions that are caused by cerebral hypoperfusion have been found to
contribute to the pathophysiological mechanisms that lead to cognitive impairment [194,195]. Animal
models of CCH showed that such compensatory actions induce abnormal activation of the frontal
cortex and the hippocampus. Hypoxemia, in addition to hypoperfusion, exacerbates ischemic brain
damage and it is associated with more severe white matter lesions. Abnormal cerebral hypoxia induces
compensatory and adaptive mechanisms to prevent hypoperfusion injury and preserve recovery of
brain function [194,196]. Part of those adaptive mechanisms involves increased capillary diameter,
neovascularization, and enhanced expression of vascular endothelial growth factor (VEGF). In the
condition of CCH, hypoxia-inducible factor 1 (HIF-1) is one of the most important transcription factors
that are involved in the endogenous adaptive response. HIF-1α then leads to the expression of a large
number of genes. It regulates more than 2% of the genes in human vascular endothelial cells [197] and is
recognized today as a regulator of the vast majority of hypoxia-inducible genes that are responsible for
the cell adaptation to hypoxia, including angiogenesis, anaerobic metabolism, mitochondrial biogenesis,
erythropoiesis, vasomotor control, and cell proliferation, such as vascular endothelial growth factor
(VEGF), glucose transporter-1 (GLUT-1), and erythropoietin (EPO), all factors that lead to survival
under hypoxic conditions [198,199]. HIF-1a is also involved in hypoxia-dependent inflammation,
apoptosis, and cellular stress. Animal models showed that the neuron-specific knockdown of HIF-1a
aggravates brain damage after a 30 min. middle cerebral artery occlusion (MCAO) and reduces the
survival rate of those mice, and an impairment of learning and memory after four weeks of CCH has
been reported. Cerebral angiogenesis is reduced, while oxidative damage is also promoted with the
proliferation of astrocytes and microglia in the cortex and some sub-regions of the hippocampus [200].
In other studies it is reported that the lowering of oxygen induces hypoxia-inducible factor-1α (that is
involved in neuroinflammatory response), which has the direct consequence of the hyper-production of
free radicals and proteases, BBB disruption, vasogenic edema, and myelin damage; all these effects may
lead to white matter (WM) damage and vascular cognitive impairment. Moreover, hypoxia-induced
MMP-9 expression leads to vascular leakage, which MMP inhibition could reduce. Pharmacological
blockage of MMP-9 or MMP-9 gene deletion confers neuroprotection in traumatic brain injury and
stroke [201].
      Protective mechanisms that are triggered by hypoxia are characterized by decreasing the O2
demand, increasing the O2 supply, or a combination of both. Some animals can reduce the O2
demand through a condition called hypometabolism, but, in the human brain, this condition is poorly
expressed. Hypoxia is always associated to early signs of failure that are represented by marked
falls in pH and tissue creatine phosphate levels, followed by a dysfunction of Na+/K+ ATPase and
lethal ion imbalance [202,203]. In the human brain, pro-survival pathways and improving brain
oxygenation actions are activated. During cerebral hypoxia, in brain, HIF-2, also known as EPAS-1
(endothelial PAS domain protein 1), is expressed, principally in endothelial cells, including brain
capillary endothelial cells [204]. HIF-2 is active during prolonged mild hypoxia and it might be involved
in brain microvascular response. In one paper, the authors provided evidence that HIF-mediated
pro-survival responses are dominant in rats with CCH. The activation of HIF-1 is part of a homeostatic
response that is aimed at coping with the deleterious effects of CCH [200]. While considering these
premises, a large number of clinical trials tried to identify protective strategies against cerebral
impairment after hypoxia through the identification of endogenous neuroprotective pathways. Based
on animal work, it has been shown that spontaneously hypertensive/stroke-prone rats (SHR/SP) with
unilateral carotid artery occlusion had white-matter damage while being treated with a permissive
Japanese diet. One week after, white matter showed a significant increase in hypoxia-inducible
Int. J. Mol. Sci. 2020, 21, 1095                                                                                          10 of 35

 factor-1α
Int. J. Mol. Sci.(HIF-1α),    which
                  2020, 21, 1095increased further by three weeks. The BBB disruption was supposed         to 35
                                                                                                      10 of  be
 secondary to hypoxia and related to a matrix metalloproteinase-9 (MMP-9)-mediated infiltration of
mediated
 leukocytes.infiltration  of leukocytes.
                In those animals,           In those
                                    treatment         animals, treatment
                                                with minocycline           with reduced
                                                                  significantly  minocycline    significantly
                                                                                          the lesion size and
reduced
 improvedthecerebral
              lesion size  andflow.
                        blood   improved   cerebralprolonged
                                     Minocycline    blood flow.survival
                                                                Minocycline
                                                                        [205].prolonged  survival
                                                                               The results are far [205].
                                                                                                   from toThebe
results
 appliedare
         in thefarhuman
                    from chronic
                           to be hypoperfusion
                                   applied in thecondition
                                                      human for
                                                              chronic   hypoperfusion
                                                                 the aforementioned      condition
                                                                                      cascades       for the
                                                                                                of events  that
aforementioned      cascades ofinevents
 appear to be determinant         humanthat
                                         SVD. appear to be determinant in human SVD.
     Insert
      InsertFigure
              Figure33approx.
                        approx.here:
                                  here:

                        Figure 3. The pathological circuit of chronic hypoxia damage in the brain.
                       Figure 3. The pathological circuit of chronic hypoxia damage in the brain.
 5. Endothelium and SVD
5. Endothelium        and SVD
        The brain endothelium,          even in severe SVD (presenting an almost complete loss of myocytes
  andThe
       other   mural    cells) remains
             brain endothelium, even      intact,  even ifSVD
                                               in severe    the endothelium
                                                                   (presenting an  is one
                                                                                      almostof the  main targets
                                                                                                 complete    loss ofofmyocytes
                                                                                                                         the redox
  altered  process   and   inflammation      (and   both  these   processes   are  highly   activated
and other mural cells) remains intact, even if the endothelium is one of the main targets of the redox  in SVD)     [134,206–209].
  This paradoxical
altered   process and  survival   of the brain
                           inflammation       (andendothelium       is also evident
                                                     both these processes             in patients
                                                                                 are highly          with CADASIL
                                                                                                activated                 [206,207].
                                                                                                            in SVD) [134,206–
  On  the  contrary,   systemic    endothelium       activation    is quite  different   in  SVD.
209]. This paradoxical survival of the brain endothelium is also evident in patients with CADASIL
        Thus,
[206,207].    Onindirectly,    brain
                   the contrary,       endothelium
                                   systemic              suffersactivation
                                                endothelium        in SVD conditions.          Mitochondrial
                                                                               is quite different    in SVD. senescence of
  the Thus,
       endothelium       walls
               indirectly,       hasendothelium
                             brain    a catastrophic      effectinon
                                                       suffers      SVD cerebral  endothelial
                                                                           conditions.             cells [210];
                                                                                          Mitochondrial           this alteration,
                                                                                                             senescence      of the
  which   is over-expressed      in  SVD    [211],  is generally    related  to  an impaired
endothelium walls has a catastrophic effect on cerebral endothelial cells [210]; this alteration, response    to the  threewhich
                                                                                                                              major
isendothelium-derived
    over-expressed in SVD      nitric  oxide-vasodilators
                                    [211],   is generally related[212], toprostacyclin
                                                                            an impaired   [213],   and endothelium-derived
                                                                                              response    to the three major
  hyperpolarizing      factors  (EDHF)     [214].  The   reduction     of NO  production      is
endothelium-derived nitric oxide-vasodilators [212], prostacyclin [213], and endothelium-derived derived  from an impairment
  of the mitochondrial      functions,   being   caused    by a  hyperproduction
hyperpolarizing factors (EDHF) [214]. The reduction of NO production is derived        of the  anti-oxidative    defensefromsystem,
                                                                                                                                 an
  and an increased
impairment       of theO2   anions reaction
                         mitochondrial          with NO,
                                            functions,      producing
                                                          being   causedperoxynitrite       [215]. The activity
                                                                            by a hyperproduction                    of endothelial
                                                                                                         of the anti-oxidative
  NO synthase
defense    system, (eNOS),
                      and an  which   catalyzes
                                increased     O2 the   production
                                                   anions   reactionofwithNO, declines    with aging
                                                                               NO, producing            [216], but is[215].
                                                                                                    peroxynitrite       even more
                                                                                                                               The
activity of endothelial NO synthase (eNOS), which catalyzes the production of NO, declineskinase
  impaired    in  SVD,   where   an  important     downstream        target  of Rho   is the   Rho-associated      protein    with
  (ROCK)
aging        [217].
        [216],  butThese
                     is evenubiquitously
                               more impaired   expressed
                                                    in SVD, serine/threonine
                                                              where an important  protein    kinases aretarget
                                                                                          downstream       involved     in diverse
                                                                                                                    of Rho   is the
Rho-associated protein kinase (ROCK) [217]. These ubiquitously expressed serine/threonine proteinof
  cellular  activities,  including     apoptosis,    smooth    muscle     contraction,    cell  adhesion,   and    remodeling
  the extracellular
kinases    are involved matrix   [218]. In
                             in diverse       the regulation
                                           cellular   activities,ofincluding
                                                                      endothelial   cell, migration
                                                                                apoptosis,      smoothROCK
                                                                                                         muscleinteracts       with
                                                                                                                     contraction,
cell adhesion, and remodeling of the extracellular matrix [218]. In the regulation of endothelial cell,
migration ROCK interacts with ezrin, radixin, and moesin (also known as the ERM proteins) that
function as cross-linkers between the plasma membrane and actin filaments [217] and are
indispensable for the leukocyte adhesion molecules coordination, being essential for barrier function
Int. J. Mol. Sci. 2020, 21, 1095                                                                   11 of 35

ezrin, radixin, and moesin (also known as the ERM proteins) that function as cross-linkers between
the plasma membrane and actin filaments [217] and are indispensable for the leukocyte adhesion
molecules coordination, being essential for barrier function [219]. Moreover, the ROCK/RhoA complex
regulates the eNOS, as previously exposed [217]. NO-induced vasodilation occurs via the activation
of myosin light chain phosphatase (MLCP) in a cGMP dependent manner. RhoA/ROCK counteracts
this through MLCP inactivation and calcium desensitization [217,220]. ROCK/Rho decreases eNOS
expression and affects the availability of NO [221]; it has also been proven in brain small vessels, even
if these effects have been largely studied in major vessel disease (coronary) [82]. Three potentially
functional eNOS polymorphisms (T-786C, intron 4ab, G894T) located toward the 50 flanking end of the
gene are known to be considered as being present in SVD and also in isolated lacunar infarction and
ischemic leukoaraiosis [222]. RhoA inhibition overwhelms VEGF-enhanced endothelial cell migration
in response to vascular injury, without, or better said, with a minimal effect, on basal endothelial cell
migration [223,224]. The maintenance of the endothelial barrier is a prior role of the endothelium
cells, mainly through the operative system of RhoA [225], also being mediated through the regulation
of Vascular endothelium cadehrins (VE-cadherins) [226]. In diabetes (one of the main risk factors
associated to SVD), advanced glycation end products (AGEs) accumulate in the vasculature, triggering
a series of purposeful and morphologic changes of endothelial cells, such as the increase of the
activation of the RhoA/ROCK pathway; the significant consequence is an increased endothelial cell
permeability [227]. It can also act as a VEGF inducer, which indirectly causes microvascular endothelial
hyper-permeability [228].
      Therefore, it should be argued that the endothelium seems to be functionally impaired in SVD,
even if morphologically and structurally undamaged [229].
      The endothelial NO downregulation in SVD is a marker of decreased endothelial regulatory
capacity, in response to external stimuli, such as hypercapnia [230,231]. Living studies have
demonstrated a significant baseline CBF reduction in SVD–affected subjects, together with an impaired
CBF autoregulation [232–234]. Endothelial activation refers to the change in the expression of
many different surface markers [235–238]. These circulating markers of endothelial activation include
intercellular adhesion molecule-1 (ICAM-1), which has been considered as a generic expression of white
matter progression [239], soluble thrombomodulin (sTM), interleukin-6 (IL-6), plasminogen activator
inhibitor-1 (PAI-1), von Willebrand factor, and others [207,240–242]. Moreover, an upregulation of
hypoxia-endothelial-related markers has been proven, such as HIF 1 alpha, VEGFR2, and neuroglobin,
when white matter lesions appear to be confluent [243]. The matter is even more impressive when
it appears evident that endothelium in overall activated, as described above, but, according to some
authors, not specifically in the human gray matter [209,241,244,245]. Though, the brain endothelium
NO dysregulation implies not only a direct inhibition of the vessel tone, but indirectly, more critically,
a decrease of the dynamic neurovascular control mechanism [246,247].
      Moreover, the permanent status of oxidative stress-induced should be taken into account, which
causes a superimposed macroscopic alteration of the cerebral endothelium.
      The immediate consequence of the endothelial dysfunction has two significant consequences, the
reduction of the resting flow in the marginally perfused white matter and macroscopic alterations
of the BBB permeability [247]; these two aspects lead to additional oxidative stress, by inducing
tissue hypoxia and extravasation of the plasma proteins [247], and both of them potentiate the
inflammation pathway, through the Nuclear Factor Kappa-Light-Chain-Enhancer of activated B cells
(NFkBeta) dependent transcription. The modern view gives the endothelium the control role of
the propagation of vasomotor signals [248], even if the question is still unresolved. In systemic
vessels, the endothelium is well known to participate in the retrograde propagation of vascular
signals [81,249], but in the brain the mechanisms by which endothelium interacts with the spread of
the vascular signal is still debated. It has been proven that a highly localized lesion of the endothelium
failed to propagate beyond the lesion site, and altered the amplitude and temporal dynamics of the
go-ahead vascular sign, with weaker temporal coordination [250]. It has been demonstrated that brain
Int. J. Mol. Sci. 2020, 21, 1095                                                                   12 of 35

endothelium is enriched with KIR channels, and not by KCa channels; these channels are sensitive to
high K flow, being derived from neural activity, and are transmitted by the synapses or by astrocytic
end-feet [249,251]. It has been recognized that K+ is recognized in the endothelium, and the upstream
penetrating arteriole is the effector of the vasodilatation [251], and its rapid propagation is probably
conducted by ionic currents traveling through the endothelium via gap junctions and then through
the myoendothelial junctions [249]. Therefore, KIR suppression avoids the increase of CBF that is
produced by cortical activation [251]. The most intriguing aspect of the endothelial conductance is
the fact that the conducted vasomotor responses, either being a dilatation, either vasoconstriction,
can be generated by different neuromodulators, i.e., Acetylcholine, ATP, prostaglandin F-2alpha, and
NO, but their effects on neurovascular coupling has never been determined [252]. The evidence is
increasing on pial arterioles: signals that are generated by the neuronal activity, deep in the brain,
should be conveyed to upstream arterioles, remote from the area of activation, to increase flow
efficiently [249]. Vascular mapping and fMRI demonstrated that vascular responses are first seen in
the deep cortical lamina during somatosensory activation, and then, more superficially, suggesting a
retrograde propagation of the vascular response [253]. A possible scenario for the transmission and
coordination of the vascular response is described [249], as follows: activation-induced increase in
extracellular potassium triggers the hyperpolarization of capillary endothelial cells and pericytes [254].
The hyperpolarization propagates upstream and reaches smooth muscle cells in penetrating arterioles,
producing relaxation [249,251]. At the same time, metabolic modifications (reduced viscosity, increased
deformability of blood cells) on the endothelium of feeding arterioles increment the smooth muscle
cell relaxation (the so-called flow-mediated vasodilation). In upstream pial arterioles, remote from the
site of activation, there is vasodilation, by propagation from arteriole downstream and acting as a local
flow-mediated and myogenic response. For all of the conditions mentioned above, SVD is defective in
neurovascular coupling, even for endothelial and pericytes failure.

6. Astrocytes and SVD
      Neurons, astrocytes, oligodendrocytes, as well as vascular and perivascular cells, are intimately
related to metabolic control and they act as trophic determinants in brain development, function,
and reaction to injury. Specifically, astrocytes play integral roles in the formation, maintenance, and
elimination of synapses in development and disease [255]. In addition to their well-established
interactions with neurons, astrocytes are also needed for the development and maintenance of BBB
characteristics in endothelial cells [256], and for the reorganization of vascular networks after brain
injury [257]. In turn, the endothelial cells regulate glycolytic metabolism in astrocytes through the
production of NO [258]. The release of vasoactive substances, such as prostanoids from astrocytes,
can couple cerebral blood flow to neuronal energy demand, and astrocytes supply neurons with vital
metabolites, such as lactate in response to neuronal activity [259]. Additional homeostatic functions
of astrocytes include water, ion, and glutamate buffering, as well as tissue repair after insult or
injury [260,261]. The reactive astrocytes can release a wide variety of extracellular molecules, including
inflammatory modulators, chemokines and cytokines, and various neurotrophic factors. These factors
can be either neuroprotective (e.g., cytokines, such as interleukin-6 [IL-6], and transforming growth
factor-b [TGF-b]) or neurotoxic (such as IL-1b and tumor necrosis factor-a [TNF-a]) [262]. The process
of glial scar formation exemplifies the interplay between the neuroprotective and neurotoxic effects of
reactive gliosis. The glial scar serves to isolate the damaged area and it prevents the damage extension
by restricting the infiltration of inflammatory cells. However, molecules that are secreted by reactive
scar-forming astrocytes can also be refractory to neurite growth [262,263]. An example of what the above
written can be found in AD, where reactive astrocytes are found in the postmortem brain of affected
patients [264–266]. It has been demonstrated that astrocytes can internalize Amyloid-beta plaques,
exerting a scavenger-like function [267,268]. Nonetheless, it has been proved that those astrocytes
with amyloid-beta are irreversibly compromised, likely showing altered calcium homeostasis [269].
Moreover, it is believed that astrocytes in AD are seriously compromised through the altered expressions
You can also read